• Non ci sono risultati.

Functional and Molecular Studies of the Crosstalk between Intestinal Microbioma and Enteric Nervous System and Potential Effects on the Gut-Brain Axis

N/A
N/A
Protected

Academic year: 2021

Condividi "Functional and Molecular Studies of the Crosstalk between Intestinal Microbioma and Enteric Nervous System and Potential Effects on the Gut-Brain Axis"

Copied!
137
0
0

Testo completo

(1)

UNIVERSITÀ DEGLI STUDI DI PADOVA

Department of Pharmaceutical and Pharmacological Sciences PhD Course in Pharmacological Sciences

Curriculum Pharmacology, Toxicology and Therapeutics

XXXI Cycle

Functional and Molecular Studies of the Crosstalk between Intestinal Microbioma and Enteric Nervous

System and Potential Effects on the Gut-Brain Axis

Coordinator: Ch.mo Prof. Piero Maestrelli

Supervisor: Ch.ma Prof.ssa Maria Cecilia Giron

PhD student: Ilaria Marsilio

2015-2018

(2)

1

Index

RIASSUNTO ... 5

ABSTRACT ... 9

1. INTRODUCTION... 13

1.1 Enteric Nervous System ... 13

1.2 Toll-like Receptors ... 19

1.2.1 Toll-like Receptors in the Nervous System ... 25

1.3 Microbiota-Gut-Brain axis ... 29

1.4 Enteric Neurotransmission ... 30

1.4.1 Cholinergic Neurotransmission ... 32

1.4.2 Tachykinergic Neurotransmission ... 33

1.4.3 Serotonergic Neurotransmission... 34

1.4.4 Nitrergic Neurotransmission... 35

1.4.5 Purinergic Neurotransmission ... 36

1.4.6 Others Neurotransmission ... 37

1.5 Oxidized Phospholipids ... 38

1.6 Intestinal Microbiota ... 41

1.7 Serotonin in the Gut and Tryptophan Metabolism ... 46

1.8 Gut Barrier and Visceral Hypersensitivity ... 49

2. AIM ... 51

3. MATERIALS and METHODS ... 53

3.1 Mice……….53

3.2 Mice Treatments ... 53

3.3 Confocal Immunohistochemistry ... 54

3.3.1 Immunohistochemistry on Frozen Sections ... 54

3.3.2 Immunohistochemistry on Ileal Whole Mount Preparations ... 55

3.3.3 Acquisition and Analysis of Images ... 57

(3)

2

3.3.4 Acetylcholinesterase and NADPH-diaphorase Biochemical Staining in Ileal

Whole Mount Preparations ... 58

3.4 In Vitro Contractility Studies ... 58

3.5 Gastrointestinal Transit Analysis ... 60

3.6 Intestinal Paracellular Permeability ... 60

3.7 Pellet Frequency and Fecal Water Content ... 61

3.8 RNA Isolation and Quantitative RT-PCR ... 61

3.9 HPLC Analysis of Tryptophan Metabolites ... 63

3.10 Statistical Analysis ... 63

3.11 Materials and Reagents ... 64

4. RESULTS ... 65

4.1 Toll-Like Receptor 4 in Murine Small Intestine ... 65

4.1.1 TLR4 Influences Ileal Morphology and ENS Architecture ... 65

4.1.2 Absence of TLR4 Impairs Gastrointestinal Motility ... 67

4.1.3 TLR4 Deficiency Affects Excitatory Neurotransmission ... 67

4.1.4 TLR4 Modulates Inhibitory Neurotransmission ... 70

4.1.5 TLR4 Absence Affects Purinergic Inhibitory Neurotransmission ... 73

4.2 TLR4 in Mouse Central Nervous System ... 74

4.2.1 TLR4 is Required for Sustaining Neuron and Glia Network in Murine Hippocampus ... 75

4.3 TLR2 and TLR4 Signaling Modulates Small Intestine Function ... 77

4.3.1 OxPAPC-mediated TLR2 and TLR4 Inhibition Alters the Architecture of the Myenteric Plexus of Juvenile Mice ... 78

4.3.2 OxPAPC Treatment Increases Excitatory Neuromuscular Contractility ... 79

4.3.3 OxPAPC Treatment Affects Inhibitory Neurotransmission ... 80

4.3.4 OxPAPC Treatment Influences Ileal SERT and 5-HT Receptors Expression.. ... 81

4.3.5 OxPAPC Treatment Modifies Serotonergic Neurotransmission ... 83

(4)

3

4.3.6 OxPAPC-mediated Inhibition of TLR2 and TLR4 Impairs Tryptophan

Metabolism... 84

4.4 Microbiota-Gut Axis Regulates Serotonergic Neurotransmission... 85

4.4.1 Antibiotic-induced Microbiota Dysbiosis Affects Visceral Sensitivity .... 85

4.4.2 Antibiotic-induced Microbiota Dysbiosis Affects Tachykinergic Neurotransmission ... 86

4.4.3 Serotonin Neurotransmission Involves Ileum Relaxation-Response following Antibiotic-induced Microbiota Dysbiosis ... 88

4.4.4 Antibiotics Treatment Influences Ileal SERT and 5-HT Receptors Expression ... 89

4.4.5 Antibiotics-induced Microbiota Dysbiosis Affects Tryptophan Metabolism... 91

4.4.6 Antibiotic-induced Microbiota Dysbiosis Causes Morphological Abnormalities in the Architecture of the Myenteric Plexus ... 93

5. DISCUSSION ... 95

6. CONCLUSIONS ... 109

7. REFERENCES ... 111

(5)

4

(6)

5

RIASSUNTO

L'interazione fra i costituenti della parete intestinale e la microflora commensale costituisce il principale artefice del mantenimento della barriera mucosale, della promozione dello sviluppo del tratto gastrointestinale (GI) e della modulazione delle funzioni GI, quali motilità, secrezione, immunità mucosale e sensibilità viscerale.

Un'alterata microflora è stata associata a disordini GI (malattia infiammatoria cronica intestinale, MICI e sindrome dell'intestino irritabile, IBS) mentre cambiamenti del microbiota intestinale durante le fasi dell’infanzia e dell’adolescenza, causati da infezioni o antibiotici, predispongono all'insorgenza di queste malattie. Inoltre, disfunzioni del sistema nervoso enterico (SNE) quali anomalie strutturali e/o variazioni nel contenuto di neurotrasmettitori, sono state associate all'insorgenza sia di MICI che di IBS. In questo contesto, giocano un ruolo chiave i recettori Toll-like (TLRs), un sofisticato sistema di proteine che attivano la risposta immunitaria innata contro agenti patogeni e mediano segnali benefici al fine di assicurare l'integrità funzionale e strutturale sia in condizioni fisiologiche che patologiche. Polimorfismi nei geni che codificano i TLRs sono stati associati a fenotipi diversi di malattia in pazienti affetti da disordini GI. In questo studio sono state caratterizzate le alterazioni strutturali e funzionali del SNE murino indotte da:

i) cambiamenti nel segnale dell’immunità innata, mediato dal recettore TLR4, ii) una miscela di fosfolipidi ossidati (OxPAPC), implicati nel blocco del segnale generato dai recettori TLR2 e TLR4 al fine di eliminare parzialmente gli effetti mediati dalla flora intestinale batterica e iii) anomalie nella composizione del microbiota.

Data l’importanza di un corretto segnale TLRs-dipendente nel mantenimento della rete

nervosa e del codice neurochimico del SNE è stata valutata la funzione intestinale in vitro

mediante esperimenti di contrattilità utilizzando la tecnica dell'organo isolato su segmenti

di ileo provenienti da topi WT e TLR4

-/-

di pari età (9 ± 1 settimane). Queste analisi hanno

evidenziato anomalie nell’attività contrattile neuromuscolare associate ad un’eccessiva

modulazione inibitoria controllata da ossido nitrico ed ATP, a sostegno della presenza di

un dialogo tra TLR4, SNE e microflora, fondamentale per la modulazione della funzione

neuromuscolare. Studi strutturali su preparati di ileo provenienti da topi TLR4

-/-

hanno

dimostrato un’alterata architettura del SNE determinata da un’anomala distribuzione

della proteina gliale strutturale GFAP (glial fibrillary acidic protein) e della subunità β

della proteina S-100, marcatore gliale nucleare e citoplasmatico in grado di legare il

(7)

6

calcio. Tali osservazioni indicano un coinvolgimento del recettore TLR4 nel mantenimento dell'integrità della rete gliale enterica mediato dalla produzione di ATP e nell’attivazione della trasmissione purinergica e pertanto evidenziano il ruolo primario di questo recettore nella conservazione dell'omeostasi strutturale e funzionale del SNE.

Inoltre, in tale modello, è stato approfondito il ruolo del recettore TLR4 nell’asse

‘intestino-cervello’ attraverso la valutazione strutturale del sistema nervoso centrale in particolare a livello dell’ippocampo, area deputata all’apprendimento. Negli animali TLR4

-/-

è stato dimostrato che la mancanza del recettore TLR4 determina nell’ippocampo, come a livello del SNE, una compromessa neuroplasticità caratterizzata da alterazioni nella densità neuronale associata a variazioni della distribuzione della rete gliale, a confermare un ruolo fondamentale del segnale TLRs anche a livello centrale.

In parallelo, è stato ulteriormente indagato il ruolo primario del segnale mediato dai recettori TLRs nell’asse microbiota-TLRs-SNE, saggiando l’effetto di una somministrazione in acuto per 3 giorni consecutivi con OxPAPC, inibitore del segnale mediato da entrambi i recettori TLR2 e TLR4, in topi adolescenti (3 ± 1 settimane). Il trattamento con OxPAPC ha causato un’alterazione significativa della risposta neuromuscolare sia recettore-mediata che non, nei topi trattati rispetto al controllo, associata a modifiche della rete neuro-gliale del SNE, confermando l’importanza del segnale mediato da tali recettori nell'assicurare l’integrità funzionale e strutturale del SNE durante l'adolescenza. Recenti studi riportano un ruolo primario nel dialogo tra i recettori TLRs e il sistema serotoninergico, spesso coinvolto in disturbi intestinali, pertanto è stato valutato se la somministrazione di OxPAPC per via intraperitoneale influenzasse tale sistema. È stato evidenziato come l’inibizione in acuto del segnale TLR2 e TLR4 comporti iperesponsività alla serotonina, alterazioni nella distribuzione recettoriale serotoninergica associata a variazioni nel metabolismo del triptofano, amminoacido coinvolto nella produzione di serotonina, a sostegno della presenza di un dialogo tra immunità innata e sistema serotoninergico.

Al fine di approfondire il ruolo dell'asse microbiota-intestino nell’omeostasi del SNE è

stato messo a punto un modello animale di deplezione di microbiota intestinale attraverso

la somministrazione intragastrica di 4 antibiotici, ampicillina (100 mg/kg), metronidazolo

(100 mg/kg), neomicina (100 mg/kg) e vancomicina (50 mg/kg) due volte al giorno per

14 giorni a topi C57BL/6J adolescenti (3 ± 1 settimane; topi ABX). Da una prima

valutazione il trattamento antibiotico ha determinato un fenotipo simil germ-free, come

già dimostrato da altri autori, ed alterazioni della motilità intestinale e dell'integrità della

(8)

7

rete neuronale e gliale enterica. A tal proposito, analisi immunoistochimiche su preparati

di ileo provenienti da topi ABX hanno evidenziato anomalie nella distribuzione ed

espressione della proteina marcatore pan-neuronale HuC/D, della proteina gliale

strutturale GFAP e della subunità β della proteina S-100. Data l’importanza di una

corretta composizione del microbiota commensale sia nel mantenimento della rete

nervosa e del codice neurochimico del SNE che nella produzione di neurotrasmettitori a

livello enterico, sono state studiate le vie di neurotrasmissione coinvolte nella sensibilità

viscerale in tale modello di disbiosi intestinale. È stato osservato un incremento dei livelli

di mRNA di GluN1 e TRPV1 nel plesso mienterico dei preparati di ileo provenienti dai

topi ABX, evidenziando gli effetti di un’alterata composizione del microbiota intestinale

sulla sensibilità viscerale. Infine, è stato valutato l’effetto di uno stato di disbiosi indotto

da antibiotici sul sistema serotoninergico, sistema le cui funzioni sono modulate da

serotonina, metabolita la cui produzione è influenzata dall’azione di specifiche spore

batteriche. Il trattamento antibiotico riporta anomalie nella risposta neuromuscolare alla

serotonina accompagnate da una compromessa rete recettoriale serotoninergica e del

metabolismo del triptofano, sottolineando l’importanza di una corretta composizione del

microbiota nel mantenimento delle funzioni mediate dal sistema serotoninergico.

(9)

8

(10)

9

ABSTRACT

The interaction between cellular constituents of gastrointestinal (GI) tract and commensal microflora is essential for the maintenance of mucosal barrier, promotion of the development of the GI system and modulation of enteric functions such as motility, secretion, mucosal immunity and visceral sensitivity. Alterations in the composition of the gut microflora have been associated to several GI disorders (e.g. inflammatory bowel disease, IBD, and irritable bowel syndrome, IBS) while changes in intestinal microbiota during infancy and adolescence, caused by infection or antibiotic therapy, appear to predispose to the onset of these diseases. Furthermore, dysfunctions of the enteric nervous system (ENS) such structural abnormalities and/or changes in the content of neurotransmitters, have been associated with the onset of IBD and IBS. In this context, a sophisticated system of proteins, so-called Toll-like receptors (TLRs), plays a key role in mediating the inflammatory response against pathogens and triggers beneficial signals to ensure tissue integrity under physiological and pathological conditions. Polymorphisms in genes encoding TLRs, including TLR2 or TLR4, have been associated with different phenotypes of disease extent and severity in patients with GI disorders. In this study we characterized structural and functional alterations of murine ENS induced by: i) changes in innate immunity response, mediated by TLR4, ii) a mixture of oxidized phospholipids (OxPAPC) that blocks both TLR2 and TLR4 signaling to partially avoid the recognition of gut commensal microflora and iii) anomalies in the composition of the microbiota.

Highlighted the role of proper TLRs signaling in the maintenance of neuronal network and neurochemical coding of the ENS, intestinal contractility was evaluated in isolated ileal segments from WT e TLR4

-/-

mice (9±1 weeks) using organ bath technique.

Functional studies reported significant alterations of intestinal contractility associated to

an increased inhibitory neurotransmission via the combined action of nitric oxide (NO)

and adenosine-5′-triphosphate (ATP), suggested a crosstalk between TLR4, ENS and

microflora in the fine-tuning of ileal contractility. Furthermore, the absence of TLR4

affects ENS architecture characterized by abnormalities in the distribution and expression

of the pan-neuronal marker HuC/D and induced a reactive gliosis state with alterations in

the glial structural protein GFAP (glial fibrillary acidic protein) and the cytoplasmatic

and nuclear glial calcium-binding protein S100β in the ileal myenteric plexus. Once

demonstrated that TLR4 signaling is involved in the control of purinergic pathways in

(11)

10

enteric neural-glial communication and highlighted its role in tuning structural and functional integrity of ENS, we assessed the role of TLR4 receptors in the central nervous system (CNS), in particular in the hippocampus assessing few architectural proteins expressed in neurons or astrocytes or microglial cells. The absence of TLR4 receptor determines neuroplasticity in the hippocampus, as well as in the ENS, characterized by a reduction of neuronal density associated with altered glial networks, to underline a key role of TLRs also in the CNS.

In parallel, to investigate the importance of TLRs-dependent signaling in modulating ENS-microbiota axis, juvenile male C57BL/6J mice (3±1 weeks old) were treated intraperitoneally with OxPAPC, that blocks both TLR2 and TLR4 signaling, twice a day for 3 days. In vivo inhibition of both TLR2 and TLR4 determined a significant alteration of receptor and non-receptor-mediated neuromuscular responses and affected myenteric plexus integrity, providing evidence that TLR2 and TLR4 signaling is essential in ensuring the structural and functional integrity of the ENS during adolescence. Recent studies demonstrated the role of TLRs in modulating intestinal serotonergic system and given that this system is involved in many GI functions, we evaluated the effect of OxPAPC treatments in this context. OxPAPC-mediated TLR2 and TLR4 inhibition affects serotonin-mediated response, in term of hyperresponsivity, and alters both serotonergic receptor distributions and tryptophan (TRP) metabolism during adolescence suggesting a cross-talk between innate immunity and serotonergic system.

To investigate the role of the microbiota-gut axis in the homeostasis of ENS we depleted gut microbiota by intragastric administration of a cocktail of broad spectrum antibiotics (50 mg/kg vancomycin, 100 mg/kg neomycin, 100 mg/kg metronidazol and 100 mg/kg ampicillin) twice a day for 14 days in adolescent mice (aged 3 ± 1 weeks, ABX). Mice after antibiotic treatment displayed a phenotype-like germ-free mice, as already reported by other Authors, and reveled an impairment in intestinal motility and in the neuro-glia integrity. Immunohistochemical analysis of ileal preparations from ABX mice showed abnormalities in the distribution and expression of the pan-neuronal marker HuC/D, the glial proteins GFAP and S100β. Given the importance of proper composition of commensal microbiota in the maintenance of neuronal network and neurochemical coding of the ENS and in the influencing neurotransmitter content, it has been investigated enteric neurotransmission involved in the control of central sensitization.

Increased mRNA levels of GluN1 and TRPV1 in the myenteric plexa of ABX mice was

found, suggesting that commensal microbiota is involved in modulating visceral

(12)

11

sensitivity. Finally, the effect of antibiotic mediated microbiota dysbiosis in serotonergic

system was evaluated. The concept of a direct communication between commensals and

the enteric nervous system was suggested by different Authors; specifically, indigenous

spore-forming bacteria from mouse and human microbiota have been shown to promote

serotonin biosynthesis. The antibiotic treatment affects serotonin-mediated response

associated with impairments of serotonergic pathways and TRP metabolism, to evidence

an involvement of microbiota in serotonin-mediated functions and potentially in

microbiota-gut axis.

(13)

12

(14)

13

1. INTRODUCTION

1.1 Enteric Nervous System

The enteric nervous system (ENS) has received special attention in the last years since it is the only limb of the peripheral nervous system (PNS) which has the ability to function independently from the central nervous system (CNS) and as such it has often been referred to as the “second brain” or the “little brain” (Goyal & Hirano, 1996).

The ENS is a complex tissue, extending from the esophagus to the anal sphincter within

the gastrointestinal (GI) system walls, and is composed of ganglia with neuronal fibers

innervating the effector tissues (Furness et al., 2014). The human ENS contains 200–600

million neurons, the same number of neurons that is found in the human spinal cord

(Furness & Costa, 1987a; Furness, 2006). The nerve-cell bodies are grouped into small

ganglia which are connected by bundles of nerve processes to form the two major

plexuses, so-called the myenteric (or Auerbach’s) plexus and the submucous (or

Meissner’s) plexus (Figure 1.1). A few small ganglia have been detected in the mucosa,

close to the muscularis mucosae (mucous plexus) (Hansen, 2003).

(15)

14

Figure 1.1. Anatomy of ENS. (A) In the small and large intestines, neurons are confined in ganglia of the myenteric plexus (MP), localized between the longitudinal (LM) and circular muscle (CM) layers, and in ganglia distributed between the circular muscle and the muscularis mucosa (MM) within the submucosa (SMuc), depicted in the transverse section of the gut wall. The ganglia and fibers in the submucosa form inner and outer submucosal plexus (SMP). (B) The distribution of ganglia along the gastrointestinal tract.

(C) Neuromuscular layers along the small and large intestines (modified from Furness, 2012).

The myenteric plexus is positioned between the outer longitudinal and inner circular muscle layers, where forms a continuous network of ganglia that extends from the upper esophagus to the internal anal sphincter (Furness, 2012). It primarily provides motor innervation to the two muscle layers and secreto-motor innervation to the mucosa. There are numerous projections from the myenteric plexus to the submucosal ganglia and to enteric ganglia of the gallbladder and pancreas (Kirchgessner & Gershon, 1990).

Moreover, a substantial number of projections from the myenteric neurons are connected to the sympathetic ganglia (Goyal & Hirano, 1996; Hansen, 2003; Figure 1.1). The myenteric plexus shows a high density of neurons compared to the submucous plexus with an average ratio of the sensory, interneurons and motor neurons of 2:1:1, respectively (Costa et al., 2000; Hansen, 2003). In large mammals, the submucous plexus is located in the submucosa and composed by an inner network located at the serosal side of the muscularis mucosae (Meissner’s plexus) and an outer layer (Schabadasch’s plexus) adjacent to the luminal side of the circular muscle layer. Moreover, in the human intestine, a third intermediate plexus lies between Meissner’s and Schabadasch’s plexus. Non- ganglionated plexuses also supply all the layers of the gut (Costa et al., 2000; Furness, 2000; Hansen, 2003). Submucosal ganglia and connecting fiber bundles form plexuses in the small and large intestines, but these ganglia are extremely rare in the stomach and esophagus (Furness, 2012; Figure 1.1).

(C)

(16)

15

The ENS is in continuous communication with autonomic nervous system (ANS) through sympathetic and parasympathetic afferent and efferent neurons. The ANS drives both afferent signals, arising from the lumen and transmitted through enteric, spinal and vagal pathways to CNS, and efferent signals from CNS to the intestinal wall. In the GI tract, sympathetic, parasympathetic, and spinal afferent nerve fibers are extrinsically innervating the ENS and ensure the bidirectional communication with the CNS through intimate connections with the spinal cord. The gut vast innervations and connections between intrinsic and extrinsic fibers guarantee the CNS monitoring of a number of gut parameters, from chemical sensing in the lumen, to sensing mechanical stress along the gut wall (Furness, 2000). Along the GI tract, the vagus nerve has three afferent endings within the gut wall: intraganglionic laminar endings within the myenteric plexus, intramuscular arrays within the smooth muscle layers and mucosal fibers within the mucosa. The stomach has the highest density of the vagal afferent ending and the density deceases towards the distal regions of the GI (Powley & Phillips, 2002). The sympathetic neurons (effector branch of the ANS) have axons that extend along the mesenteric nerves deep into the gut wall to the myenteric, submucosal and mucosal plexuses of the ENS (Lomax et al., 2010). The terminals of these axons are responsible of releasing numerous neurotransmitters, mainly norepinephrine (NE) and tyrosine hydroxylase (TH). In the other side, vagal efferent neurons of the motor pathways are parasympathetic preganglionic neurons (Hansen, 2003). A variety of central effects, primarily on the upper GI tract, are mediated through these neurons, including relaxation of the proximal stomach, enhancement of gastric peristalsis, and promotion of gastrin secretion.

Transmission from vagal input neurons to enteric neurons is mediated principally by acetylcholine (ACh) acting on nicotinic cholinergic receptors, but several other transmitters are involved in these processes (Hansen, 2003). This bidirectional connection, the so-called gut-brain axis, provides neural control of all functions of the GI tract (Goyal & Hirano, 1996). The ENS is endowed with a wide array of restorative, maintenance and adaptive functions. Motility patterns, gastric secretion, transport of fluid across the epithelium, blood flow, nutrient handling, interaction with the immune and endocrine systems of the gut are function under the control of the ENS (Furness, 2012;

Wood, 2012).

According to neurons morphology, neurochemical coding, cell physiology, projections to

targets and functional roles, approximately 20 distinct types of neurons have been

(17)

16

described (Costa et al., 2000). The enteric neuronal circuits are composed by intrinsic primary afferents neurons, sensory neurons which detect mechanical distortion of the mucosa, mechanical forces in the external musculature (tension of the gut wall) or the presence of chemical luminal stimuli and initiate appropriate reflex control of functions including motility, secretion and blood flow (Clerc et al., 2002). Along the whole GI tract, the longitudinal and circular smooth muscle layers and the muscularis mucosae are innervated by uni-axonal excitatory and inhibitory motor neurons (Dogiel type I morphology), which receive prominent fast excitatory synaptic potentials (Wood, 2012).

The primary neurotransmitters for excitatory motor neurons are ACh and tachykinins.

Several neurotransmitters have been identified in inhibitory motor neurons, including nitric oxide (NO), vasoactive intestinal peptide (VIP) and adenosine triphosphate (ATP)- like transmitters, although NO is considered the primary transmitter (Furness et al., 2014;

Table 1.1).

Table 1.1. Proportions of all neurons attributed to different functional classes in myenteric ganglia of mouse small intestine (modified from Qu et al., 2008; Hao et al., 2013). Abbreviations: AH, after-hyperpolarizing;

S, synaptic.

(18)

17

Another important class of enteric neurons is represented by secretomotor and secretomotor/vasodilator neurons regulating the electrolyte and water transport across the intestinal mucosa (Vanner & Macnaughton, 2004).

In the ENS, in parallel with neuron population, it is possible to identify another cell population that is represent by enteric glial cells (EGCs). In the last couple of years, the role of EGCs in ENS function has gained significant attention (Sharkey, 2015). EGCs constitute a major population of peripheral glia that is located within the ganglia of the myenteric and submucosal plexus of the ENS and in extraganglionic sites, such as the smooth muscle layers and the mucosa (Gershon & Rothman, 1991; Gulbransen &

Sharkey, 2012; Ruhl et al., 2004). The EGCs are usually small cells with highly irregular, stellate-shaped body, associated to neuronal cell bodies in enteric ganglia in an intimate physical connection, highly reminiscent of the relationship between astrocytes and neurons in the CNS (Gulbransen & Sharkey, 2012; Figure 1.2). EGCs also show connections with enteric nerve fiber bundles, which are similar to peripheral Schwann cells, but differ from these by the function (Lomax et al., 2005). Different types of EGCs have been identified (Boesmans et al., 2015; Hanani & Reichenbach, 1994) and are subdivided into four subtypes which correspond to unique locations within the plexus and extraganglionic spaces and to specific phenotypic properties (Boesmans et al., 2015). The EGCs ‘type I’ or ‘protoplasmic’ display star-shaped cells with short, irregularly branched processes resembling protoplasmic astrocytes of the CNS and closely embrace neuronal cell bodies and fibers within myenteric and submucosal ganglia (intraganglionic EGCs).

Enteric glia ‘type II’ represents the elongated glial cells within interganglionic fiber tracts, which are similar to fibrous astrocytes of the white matter in the CNS. The subepithelial glia consists of several long branches that reach the mucosal epithelial cells, and thus could be grouped as ‘mucosal’ or ‘type III’ EGCs. The fourth type of enteric gliocytes are distributed between smooth muscle cells, running with neuronal fibers in the musculature, thus these cells are ‘intramuscular’ or ‘type IV’ EGCs (Hanani &

Reichenbach, 1994; Figure 1.2). Traditionally, EGCs were thought to contribute

primarily to the structural integrity and nourishment of the ENS. However, during the last

decades several studies have confuted the concept of a merely supportive function of

EGCs and ascribed to them a wide variety of roles that are essential for proper GI function

(Boesmans et al., 2015). In addition to be a scaffold for neurons, EGCs are involved in

most gut functions such as mucosal integrity, neuroprotection, adult neurogenesis, neuro‐

(19)

18

immune interactions, and synaptic transmission (De Giorgio et al., 2012; Gulbransen &

Sharkey, 2012; Neunlist et al., 2013; Ruhl et al., 2004).

Figure 1.2. Subpopulations of enteric glia. (A) Several subpopulations of enteric glia located within the gut wall with different proposed physiological functions and signaling mechanisms. (B) Mucosal enteric glia lies in the mucosa directly beneath the epithelial cells. (C) Intraganglionic glia surround neurons (blue) within the enteric nerve plexuses (submucosal and myenteric plexus). (D) Intramuscular glia is associated with enteric nerve fibers innervating the smooth muscle layers (circular muscle and longitudinal muscle).

Abbreviations: α2-AR, α2 adrenergic receptor; 15d-PGJ2, 15-deoxy-Δ12,14-prostaglandin J2; GAT2, sodium- and chloride-dependent GABA transporter 2; mGluR5, metabotropic glutamate receptor 5;

NTPdase2, ectonucleoside triphosphate diphosphohydrolase 2; PAR1/2, protease-activated receptor 1/2;

PEPT2, peptide transporter 2 (also known as solute carrier family 15 member 2); proEGF, proepidermal growth factor; P2X7, P2X7 receptor; P2Y1,2,4, P2Y1,2,4 receptor; TGF-β, transforming growth factor β.

(modified from Gulbransen & Sharkey, 2012).

Although at present EGCs are the least-studied peripheral glial cells in mammals, there

is an increasing interest in understanding the complex roles of these cells in GI

physiology. Clonal cultures of ENS progenitors have shown that EGCs originate from

common neuro-glial progenitors (Bondurand et al., 2003), but the presence of bi-potential

or committed neurogenic and gliogenic progenitors in vivo has not been documented so

far. Moreover, it remains unclear the role of individual progenitors in generating distinct

subtypes of enteric neurons and glial cells. In rodents, a significant fraction of enteric

neurons and EGCs develops during the early postnatal period, and it would be very

interesting to explore how changes associated with feeding or the establishment of

luminal microflora and the maturation of the mucosal immune system after birth affect

(20)

19

ENS development (Kabouridis & Pachnis, 2015). Recently, emerging evidence suggests that gut microflora can have dramatic effects on the development and function of the nervous system, both at the local as well as at the systemic level (Obata & Pachnis, 2016).

The role of microbiota on ENS organization is highlighted by the reduced number of enteric neurons and the associated deficits in gut motility observed in germ-free (GF) mice (Anitha et al., 2013). Furthermore, the development and continuous homeostatic influx of EGCs into the intestinal mucosa is defective in GF mice or in antibiotic-treated mice (Kabouridis et al., 2015). These findings reveal the complex and intricate relationship between the microbiota and EGCs as regulators of neuroimmune control of host defense in the intestinal mucosa (Sharkey et al., 2018) and essential for the assembly of intestinal neural-glial circuits. Interestingly, reconstitution of GF mice with conventional microbiota normalized the density of EGCs network and gut physiology (Kashyap et al., 2013; Kabouridis et al., 2015) raising interesting questions relating to the cellular plasticity of the ENS and the mechanisms by which microbiota influence its homeostasis. Furthermore, the potential role of the microbiota and the mucosal immune system in the activation of glial progenitors and the homeostasis of EGCs is currently unclear, but it is interesting that glial cells are capable to direct influence immune responses (Turco et al., 2014). To this concern, an upon bacterial stimulation, EGCs upregulate expression of MHC class II, which suggests that they actively respond to the colonization of the gut lumen by microbiota and participate in antigen presentation to the adaptive immune system (Turco et al., 2014). Taken together, these observations highlight that dynamic host–microbe interactions are a key element for EGCs development, suggesting that an improved understanding of this mechanism will provide important insights into the pathophysiology of GI diseases.

1.2 Toll-like Receptors

All living organisms are constantly exposed to environmental microorganisms and cope

with their potential invasion into the body. The vertebrate immune response can be

divided into innate and acquired immunity. The innate immune system is the first line of

host defense against pathogens and is mediated by phagocytes including macrophages

and dendritic cells (DCs; Akira et al., 2006). In fact, to control the infection during the

first days, the organism, through innate immune system, modulates some important

functions including opsonization, activation of complement, coagulation cascades,

(21)

20

phagocytosis, activation of proinflammatory signaling cascades and apoptosis (Janssens

& Beyaert, 2003). By contrast, acquired immune responses are slower processes, in the late phase of infection, which are mediated by T and B cells, both of which express highly diverse antigen receptors that are generated through DNA rearrangement and are thereby able to respond to a wide range of potential antigens and to generation of immunological memory (Akira et al., 2006). This highly sophisticated system of antigen detection is found only in vertebrates and has been the subject of considerable research. Far less attention has been directed towards innate immunity, as it has been regarded as a relatively nonspecific system, however is able to discriminate between self and non-self, such as a variety of pathogens and to present antigen to the cells involved in acquired immunity (Akira et al., 2006). Also, the innate immune system has an important function in activation and shaping of the adaptive immune response through the induction and release of co-stimulatory molecules and cytokines (Medzhitov, 2007; Figure 1.3). In contrast to the clonotypic receptors, expressed by B and T lymphocytes, the innate immune system uses nonclonal sets of recognition molecules, called pattern recognition receptors (PRRs; Janssens & Beyaert, 2003; Figure 1.3).

Figure 1.3. Pathways of host-defense mechanisms (modified from Medzhitov, 2007). Abbreviation: PRRs, pattern recognition receptors.

Toll like receptors (TLRs) are one of the most important family of the PRRs. The

(22)

21

discovery of the TLRs started with the identification of the receptor ‘Toll’, a protein expressed in Drosophila melanogaster and involved in controlling embryonic development (Akira & Takeda, 2004; Okun et al., 2011). Subsequent genetic studies have led to the discovery of genes important in the dorsal-ventral patterning of the embryo (i.e., the dorsal group of genes, including Toll, tube, pelle, cactus, the NF-κB homolog dorsal, and seven genes upstream of Toll; Belvin & Anderson, 1996). Since NF-κB is involved in mammalian immunity, gradually became evident the contribution of TLRs in the signaling pathways in regulating Drosophila embryonic development and activating the immune system (Wasserman, 1993). In the 1995, Hultmark and colleagues first identified Toll-1 as an activator of the immune response in a Drosophila cell line. Around the same time, a human homolog of Toll was identified and mapped to chromosome 4p14 (Taguchi et al., 1996). Later on, an in vivo study in Drosophila demonstrated that the Toll signaling is involved also in the antifungal response (Lemaitre et al., 1996). In the 1997, the first mammalian TLRs was described by the group of Medzhitov. Subsequently, five human TLRs have been characterized (Rock et al., 1998) that are involved only in controlling immune responses with no role in the development whereas the Drosophila Toll pathway is implicated both in immunity and developmental processes (Valanne et al., 2011). TLRs are type I transmembrane proteins responsible in the recognition of foreign pathogens referred to as pathogen-associated molecular patterns (PAMPs). PAMPs are well suited to innate immune recognition for three main reasons: i) they are invariant among microorganisms of a given class; ii) they are products of pathways that are unique to microorganisms, allowing discrimination between self and non-self-molecules; iii) they have essential roles in microbial physiology, limiting the ability of the microorganisms to evade innate immune recognition through adaptive evolution of these molecules (Medzhitov, 2007). Bacterial PAMPs are often components of the cell wall, such as lipopolysaccharide (LPS), peptidoglycan (PG), lipoteichoic acids (LTA) and cell-wall lipoproteins. An important fungal PAMP is beta-glucan, which is a component of fungal cell walls, but also viral nucleic acids structures are recognized by TLRs. An important aspect of pattern recognition is that PRRs themselves do not distinguish between pathogenic microorganisms and symbiotic (non-pathogenic) microorganisms, because the receptor ligands are not unique to pathogens (Medzhitov, 2007). So far, 10 and 12 functional TLRs have been identified in humans and mice, respectively, with TLR1–

TLR9 being conserved in both species. Mouse TLR10 is not functional for a retrovirus

insertion, and TLR11, TLR12 and TLR13 have been lost from the human genome (Kawai

(23)

22

& Akira, 2010; Table 1.2). Studies in mice deficient in each single TLRs type have demonstrated that every TLR has a distinct function in terms of PAMPs recognition and activation of immune responses (Akira et al., 2006).

TLR1, 2, 4 and 6 recognize lipid-based structures. TLR4 recognizes LPS from Gram- negative bacteria, which causes septic shock (Akira et al., 2006). TLR2 forms heterodimers with TLR1 and TLR6 and in concert with TLR1 or TLR6 discriminates between the molecular patterns of triacyl and diacyl lipopeptide, respectively, which derived from Gram-positive bacteria, mycoplasma and mycobacteria (Kumar et al., 2009). TLR5 and 11 recognize protein ligands. TLR5 is expressed abundantly in intestinal CD11c-positive lamina propria cells where it senses bacterial flagellin (Uematsu &

Akira, 2006). TLR3, 7, 8 and 9, being localized intracellularly, detect nucleic acids

derived from viruses and bacteria. TLR3 was shown to recognize double stranded RNA

(dsRNA) generally produced by many viruses during replication. TLR7 recognizes

synthetic imidazoquinoline-like molecules, guanosine analogs such as loxoribine, single

stranded RNA (ssRNA) derived from viruses and small interfering RNA (Akira et al.,

2006; Table 1.2). TLRs are expressed on a variety of cells, including immune cells, such

as macrophages, DCs, B cells, specific types of T cells, and also fibroblasts, epithelial

cells and neurons. Expression of TLRs is not static but rather is modulated rapidly in

response to pathogens, an array of cytokines and environmental stressors (Akira et al.,

2006). Furthermore, TLRs may be expressed extracellularly or intracellularly. While

certain TLRs (TLRs 1, 2, 4, 5, and 6) are expressed on the cell surface, others (TLRs 3,

7, 8, and 9) are found almost exclusively in intracellular compartments such as

endosomes, and their ligands, mainly nucleic acids, require internalization to the

endosome before receptor signaling is possible (Akira et al., 2006).

(24)

23

Table 1.2. Descriptions of TLR location and characteristics (modified from Kumar et al., 2009; Duffy &

O'Reilly, 2016).

The engagement of TLRs by microbial components triggers the activation of signaling cascades, leading to the induction of genes involved in antimicrobial host defense. TLRs are characterized by an ectodomain composed of leucine rich repeats (LRR) that are responsible for recognition of PAMPs and a cytoplasmic domain homologous to the cytoplasmic region of the IL-1 receptor, known as the TIR domain, which is required for downstream signaling (Kawai & Akira, 2007).

After ligand binding, TLRs dimerize and undergo conformational changes required for

the recruitment of TIR-domain-containing adaptor molecules of the TLR (Akira et al.,

2006). The adaptor molecules include myeloid differentiation factor 88 (MyD88), TIR-

associated protein (TIRAP)/MyD88-adaptor-like (MAL), TIR-domain-containing

adaptor protein-inducing IFN-b (TRIF)/TIR-domain-containing molecule 1 (TICAM1)

(25)

24

and TRIF-related adaptor molecule (TRAM; Oshiumi et al., 2003; Yamamoto et al., 2002;

Figure 1.4).

Figure 1.4. TLR signaling in conventional dendritic cells, macrophages and plasmatic dendritic cells.

Abbreviations: IKK complex, Inhibitor of nuclear factor kappa-B kinase complex; IKKα, Inhibitor of nuclear factor kappa-B kinase subunit alpha; IRAK4,1,2, Interleukin-1 receptor-associated kinase 4, 1, 2;

IRF3, 7, Interferon regulatory factor 3, 7; MAP kinase, Mitogen-activated protein kinase; MyD88, myeloid differentiation factor 88; NFkB, Nuclear-factor kappa B; RIP1, receptor-interacting protein 1; TLR, Toll- like receptor; TAK1, Transforming growth factor beta-activated kinase 1; TBK1/KKi, kinase binding domain; TIRAP, Toll/interleukin-1 receptor domain-containing adapter protein; TRAM, Translocating chain-associated membrane protein 1; TRAF3,6, TNF receptor-associated factor 3; TRIF, TIR-domain- containing adapter-inducing interferon-β (modified from Kumar et al., 2009).

The differential responses mediated by distinct TLRs ligands can be explained in part by

the selective usage of these adaptor molecules. MyD88 and TRIF are responsible for the

activation of distinct signaling pathways, leading to the production of pro-inflammatory

cytokines and type I IFNs, respectively (Kumar et al., 2009). MyD88 is a universal

adapter that activates inflammatory pathways; it is shared by all TLRs with the exception

of TLR3. For the complexity of the pathway, the TLRs signaling pathway is categorized

into MyD88-dependent and TRIF-dependent pathways (Akira et al., 2006). Upon

stimulation, MyD88 associated with the portion of TLRs recruits IL-1R associated kinase

(IRAK), which leads to the activation of TNF receptor associated factor 6 (TRAF6) to

promote stimulation of TAK1 which results in the activation of MAP kinase or NF- κB

through IKK complex, resulting in the induction of genes involved in inflammatory

(26)

25

response (Akira et al., 2006). Also, TIRAP mediates the activation of MyD88-dependent pathway. While TRIF activates an alternative pathway to induce a production of inflammatory cytokines and type I interferons (INFs). TRIF interacts with receptor- interacting protein 1 (RIP1), through a MyD88-independent way, determining the production of several cytokines (Kawai & Akira, 2007). The TRIF-dependent pathway induces INFs through IRF3 that is phosphorylated and activated by IKK-related kinase (TBK1 and IKKi) via TRAF3, a linker between TRIF and TBK1 (Kumar et al., 2009).

TLR9 and TLR7 mediated INFs secretion in a MyD88-dependent manner, in contrast to TLR3 and TLR4 that produce TRIF-dependent IFN response (Kumar et al., 2009; Figure 1.4).

1.2.1 Toll-like Receptors in the Nervous System

In the gut, resident bacteria confer many benefits to intestinal physiology and have a truly mutualistic relationship with the host (Hooper & Gordon, 2001). However, inappropriate activation of the immune system by commensal bacteria or pathogens appears to play crucial importance in the pathogenesis of intestinal disease (Podolsky, 2002).

Intriguingly, GF animal studies demonstrate that the microbiota is necessary for the

development of gut mucosal immunity (Macpherson & Harris, 2004). TLRs play a role

in the cross-talk between the intestinal microbiota and the host, as they specifically

recognize conserved microbial molecular structures, called MAMPs (Martin et al., 2010)

and protect against pathogenic microorganisms. Furthermore, microbiota-driven immune

response can prevent the development of inappropriate inflammatory response to

commensal microbiota and establish a host-microbial homeostasis. A breakdown of this

gut homeostasis due to microbial dysbiosis could cause immune-related disorders

(Manichanh et al., 2006), diabetes (Wen et al., 2008), allergies (Penders et al., 2007) and

obesity (Ley et al., 2005). The composition of intestinal microbiota has an important role

in shaping host immunity, including cytokine expression, development of GALT and

mucosal barrier. GF mice have numerous immune abnormalities, including failure of

secondary lymphoid development, lower levels of antimicrobial peptides and smaller

numbers of intraepithelial lymphocytes (Sauza et al., 2004). Colonization of GF mice can

restore the proper organization of the intestinal immune system (Hooper, 2004). Thus,

microbiota can enhance innate immunity through mucous secretion and production of

antimicrobial peptides. These data show that the precise composition of the intestinal

(27)

26

microbiota can qualitatively and quantitatively influence host immune responses, which in turn have the capacity to affect gut function.

The expression of TLRs in both CNS and ENS (Barajon et al., 2009) has suggested that TLRs are not only involved in regulating host immune responses but, also, they may have a role in central aspects of neuroinflammation, neurodevelopment and neuroplasticity (Aravalli et al., 2007; Okun et al., 2011). Among all TLRs the most important bacteria- sensor proteins are TLR2 and TLR4; since they are expressed by enteric neurons and glia, which suggest that ENS lineages can directly sense microbial microbiota. TLR2 recognizes large variety of PAMPs, in particular different ligand such as porins, lipoprotein, LTA, bacterial PG, viral hemagglutinin and glycoproteins (component of Gram-positive bacteria) and their interaction leads to the activation of MyD88-dependent signaling pathways (Takeda et al., 2003). The importance of TLR2 in the host defense against Gram-positive bacteria has been demonstrated using TLR2-deficient (TLR2

-/-

) mice, which have been found to be highly susceptible to challenge with Staphylococcus aureus or Streptococcus pneumoniae (Takeuchi et al., 2000; Echchannaoui et al., 2002).

TLR2 appears also to have a crucial role in host defense against extracellular growing of Gram-positive bacteria (Akira & Takeda, 2004). On the other hand, TLR4 has been found to detect LPS, a major component of Gram-negative bacteria cell wall (Takeda et al., 2003). The stimulation of TLR4 by LPS results in the activation of MyD88-dependent and MyD88-independent pathways, leading to the production of several inflammatory cytokines and IFN-beta associated with the expression of IFN-inducible genes, respectively (Akira & Takeda, 2004). Several studies advocate for a role of TLRs in ENS homeostasis (Kabouridis & Pachnis, 2015). Recently TLR2 and TLR4 signaling seems to be fundamental for ensuring intestinal integrity and protecting from harmful injuries, in fact changes in their expression have been reported in functional and/or inflammatory bowel disease (IBD, Rakoff-Nahoum et al., 2006). Changes in the architecture and neurochemical coding of ENS lead to gut dysmotility and to higher IBD susceptibility in a model of TLR2

-/-

mice highlighting TLR2 as major player in gut homeostasis (Brun et al., 2013). Furthermore, myenteric ganglia of TLR2

-/-

mice contained fewer neurons compared with their wild-type mice, with reduction in inhibitory nNOS

+

neurons being the most notable phenotype (Brun et al., 2013). The reduction in nNOS

+

neurons is accompanied by intestinal dysmotility and impaired chloride secretion in ileum.

Administration of GDNF can correct many of the ENS deficiencies in TLR2

-/-

mice and

(28)

27

in antibiotic-treated animals, suggesting that one of the roles of the microbiota-TLR2 axis is to promote the expression of neurotrophic factors that are required to maintain the functional organization of the mammalian ENS (Brun et al., 2013). Some studies reported also, that the absence of TLR2 increases susceptibility to intestinal injury and inflammation (Cario et al., 2007). Anitha, and colleagues, showed that GF and antibiotic- treated mice exhibited reduced motility and fewer nNOS

+

neurons (Anitha et al., 2012).

This effect was mediated, at least partly, via TLR4, since TLR4

-/-

mice exhibited similar deficits in intestinal motility and a reduced number of nitrergic neurons as GF mice. This phenotype was reproduced in mice with ENS specific MyD88 knockout, suggesting that TLR4 signaling is critical for the nitrergic neurons within ENS lineages. The same study demonstrated that LPS promoted the survival of cultured enteric neurons in an NF-κB–

dependent manner (Anitha et al., 2012). TLR4 is the best characterized pathogen- recognition receptor and recently recognized to modulate ENS phenotype and function (Anitha et al., 2012; Caputi et al., 2017a). Our group recently demonstrated the role of TLR4 in controlling small bowel contractility through nitrergic-purinergic neurotransmission (Caputi et al., 2017a) and in modulating the distribution of EGCs in the ileum and the concomitant release of two signaling molecules, NO and ATP involved in controlling GI motility (Caputi et al., 2017a). Thus, the cross-talk between TLR4 and nitrergic/purinergic pathways in neural-glial communication is likely to be a prerequisite for understanding normal gut physiology and the pathology. Polymorphisms in TLRs genes or in general a defective immune response, appear to be involved in the initiation and perpetuation of chronic inflammation in IBD (Pierik et al., 2006).

Apart from PAMPs/MAMPs-derived ligands, TLRs also sense endogenous molecules released from stressed or dying cells—termed damage- or DAMPs, mainly derived from tissue damaged by oxidative stress. For example, TLR4 recognizes heat shock protein (Hsp) 60, Hsp 70 and fibrinogen and TLR2 recognizes Hsp 70, hyaluronan, and versican (Kim et al., 2009). After recognition of DAMPs, TLRs activate and orchestrate several innate immune machineries, promoting apoptosis and shaping adaptive immune responses, but the deregulation of this response can lead to inflammatory collateral tissue damage and some forms of autoimmunity and autoinflammatory diseases (Land, 2015).

TLRs are also expressed in several residing cells of the CNS such as astrocytes, microglia,

oligodendrocytes and neurons, and the regulation of their expression seems to be dynamic

and associated with profoundly changes during aging (Letiembre et al., 2007). In the

(29)

28

brain, TLRs can be activated not only after the invasion of pathogens but also in the absence of microbial infection (Zhang & Schluesener, 2006) and regulate neurogenesis through the release of growth factors (Rolls et al., 2007). Okun and colleagues suggested the existence of a paradigm in which exists an auto-regulation of the innate immune system in the CNS, which helps to prevent excessive inflammation during pathogen infections (Okun et al., 2009). Several authors have recently highlighted the involvement of TLR4 in CNS plasticity, learning and memory, and behavior such as novelty seeking and social interaction (Okun et al., 2012; Li et al., 2016). Zhu et al (2016) showed various cerebellum-related motor defects in TLR4-deficient mice, due to the loss of Purkinje cells. Furthermore, TLR4 is involved in modulating the self-renewal and the cell-fate of neuronal stem/progenitor cells (Rolls et al., 2007). These findings suggest that TLR4 signaling is essential for neuron development and plasticity in the CNS. Other “pivotal”

players in the innate regulation of inflammatory responses in the CNS are microglial cells,

once activated by inflammatory stimuli, operate to maintain CNS integrity. However, in

case of massive and uncontrolled release of proinflammatory mediators, microglia may

cause severe neuronal damage (Aravalli et al., 2007). Also, it should be noted that changes

in the permeability of blood-brain barrier (BBB) are crucial for the infiltration of

antibodies and lymphocytes from peripheral tissues, leading to the degeneration of the

neuronal structure (Nguyen et al., 2002). In this case the condition is critical, because

alterations of BBB are possibly linked to increased vulnerability of CNS cells and

excessive innate immune responses together with the production of cytokines, a condition

known as excitotoxicity (Nguyen et al., 2002). Alternatively, a peripheral challenge can

generate a systemic inflammation with the secretion of molecules of innate immune

system that are able to cross the BBB and damage the CNS (Yang et al., 2000). In fact,

some authors demonstrated that perturbed stability of the BBB is present in

neurodegenerative disease such as Alzheimer’s disease, stroke and amyotrophic lateral

sclerosis (Huber et al., 2001). This instability of the barrier is associated with a severe

inflammation and overexpression of TLRs, and the dynamic expression of these receptors

seem to be involved in the progression of neurodegenerative pathologies in both normal

aging and age-related disease (Okun et al., 2009).

(30)

29

1.3 Microbiota-Gut-Brain axis

In the last decade, emerging evidence has revealed the presence of an intense dialogue between the brain and the GI system, the so-called brain-gut axis, and furthermore that microbiota can influence not only the immune and metabolic systems, but also the nervous system (Collins et al., 2012). The gut–brain axis is pivotal in maintaining homeostasis and is involved in the control of diverse physiological functions including motor, sensory, autonomic, and secretory functions of the GI tract to regulate an array of processes from energy metabolism to mood regulation (Burokas et al., 2015; Dinan et al., 2015). Communication between CNS and ENS implies a bidirectional connection system:

the brain influences the function of the ENS whereas the gut influences the brain via vagal and sympathetic afferents. The ENS independently controls gut function, the migrating motor complex, and peristalsis, but it is constantly monitored and modified by CNS via both vagal and sympathetic extrinsic nerves. Lately, it is becoming increasingly clear that a third player, such as the gut microbiota, can significantly influence the gut-brain crosstalk, having a marked impact on digestive processes, immune responses, emotional status, perception and cognitive functions (Felice et al., 2016). The microbiota-gut-brain axis has attracted much attention regarding the pathogenesis of different central neurodegenerative disease, in which GI dysfunction appears many years before of degenerative state. In addition, the enteric microbiota is a huge antigenic load resident in the gut and confers marked potential danger if not kept under continuous surveillance, such as under TLRs sensing (Caputi & Giron, 2018). However, the enteric commensal microbiota is required for the constant stimulation of the immune system and TLR- mediated sensing of these microorganisms may play a dual role in disease development as a source of both inflammatory and regulatory signals. In this respect, it is important to take into account that the microbiota is also a source of biological active signaling molecules, immune mediators and gut hormones. Some of those, including serotonin, purines, GABA and neurotrophic factors, among others, have been shown to be involved in TLRs signaling (Brun et al., 2013, 2015; Latorre et al., 2016; Caputi et al 2017a, b).

The emerging evidence described above suggests the existence of a multifaceted TLR

signaling network that influences neural circuits and immune-mediated processes both in

the gut and in the brain. Further studies focused on discovering the enteric microbiota-

derived factors responsible of TLRs engagement and the consequent signaling outcomes

of TLR activation in both the ENS and the CNS will provide novel insights into the

(31)

30

complex dialogue between the host and the microbiota in neurodegenerative disorders (Caputi & Giron, 2018).

1.4 Enteric Neurotransmission

Neural networks for the control of digestive functions are positioned at many levels: the CNS, spinal cord, prevertebral sympathetic ganglia, and in the wall of the specialized tissues that composed the digestive system. The major actors of this system are the neurons, which usually express a combination of different neurotransmitters, a phenomenon known as chemical coding. The chemical code depends on the type of neuron, the species and the intestinal segment (Hansen, 2003). The resting membrane potential of enteric neurons is normally less negative than in the CNS (-40 to -70 mV) and is largely determined by potassium channels. Hyperpolarization induces inhibition, whereas depolarization induces excitation of the neuron (Furness et al., 2000). Action potentials are mostly carried by sodium (extrinsic nerves) and calcium (intrinsic nerves) (Vanden Berghe et al., 2001). The general mechanism of chemically mediated synaptic transmission in the ENS is complex as in the CNS. More than 30 neurotransmitters have been identified in the ENS (Galligan, 1998; Furness et al., 2000). Enteric neurotransmitters are either small molecules (e.g. norepinephrine and serotonin), larger molecules (peptides) or gases (NO and carbon monoxide).

The last classification of enteric neurons relies on their functional proprieties and three types of neurons can be distinguished.

✓ SENSORY NEURONS: these neurons are divided into two categories:

▪ Intrinsic primary afferent neurons (IPANs) belong to AH type (AFTER HYPERPOLARISATION), present an oval or rounded soma and their prolongations contact different neurons in the mucosal and sub-mucosal plexus. Their activation after mechanical or chemical stimuli applied to the intestinal mucosa allow the ENS to generate appropriate reflex responses.

Chemosensitive IPANs respond to chemical stimuli applied on the luminal

surface of the small intestine’s mucosa while mechanoreceptor myenteric

IPANs react to intestinal wall musculature’s contraction through

mechanosensitive ion channels (Kunze et al., 2000). Sub-mucosal IPANs

(32)

31

instead response indirectly to mucosal distortions through enterochromaffin cells that release serotonin after mechanical stimulus.

▪ Extrinsic primary afferent neurons (EPANs) can be vagal or spinal afferents where the first involved in physiological events while the second primary response to physiological stimuli.

Sensory neurons can be also nociceptors because their activation after a nociceptive stimulus evokes protective responses (Furness et al., 2014).

✓ INTERNEURONS: usually these neurons belong to Dogiel type II and could be S- (synaptic) or AH type. They are interposed between the primary afferent neurons and the motor or secretomotor neurons. Four types of interneurons constitute chains extended along the intestine: one ascendant interneuron is cholinergic and is the conduct for the ascendant path that compose the projectile reflexes. Three descendant interneurons have a complex chemical distinction and studies performed on the connections established by neurons revealed that each type perform different actions (Costa et al., 2000).

▪ ChAT (choline acetyltransferase)/NOS (nitric oxide synthase)/VIP (vasoactive intestinal peptide) interneurons are involved in local motor reflexes (Costa et al., 2000).

▪ ChAT/SOM (somatostatin) interneurons play a key role in the conduction of migrating myoelectric complexes in the small intestine (Costa et al., 2000).

▪ ChAT/5HT (serotonin) interneurons are associated in secreto-motor but not directly in motor reflexes (Costa et al., 2000).

✓ MOTONEURONS: these neurons belong to Dogiel type I and S-type (Hansen, 2003).

▪ Muscular motoneurons innervate the muscularis mucosae, longitudinal and circular musculature of the entire digestive tract. They mediate cholinergic and tachykinergic excitatory stimulation but also inhibitory arousal (Hansen, 2003).

▪ Secretomotor and vasomotor neurons control secretion and blood flow and

are directly regulated by IPANs through the release of ACh and VIP. The

soma of the majority of secretomotor and vasomotor neurons is in the sub-

mucosal plexus and some of these neurons project in the myenteric plexus

(33)

32

while other in the muscularis mucosae. These neurons can be either cholinergic or non-cholinergic: ACh released by cholinergic neurons acts on muscarinic receptors in the mucosal epithelium, VIP instead is released by non-cholinergic neurons as transmitter (Hansen, 2003). Sympathetic afferents modulate local reflexes loop that regulate secretion and blood flow (Hansen, 2003).

1.4.1 Cholinergic Neurotransmission

The major excitatory transmission within the ENS are mediated by cholinergic transmission, with ACh, producing excitatory potentials in post-synaptic effectors. The cholinergic circuitry of the ENS is extensive and mediates motility (muscular) and secretory (mucosal) reflexes, in addition to intrinsic sensory and vascular reflexes. ACh is a co-transmitter of the greater population of the enteric neurons and is synthesized in nerve terminals from choline and acetyl-CoA by choline acetyltransferase (ChAT) and is then translocated to synaptic vesicles by the vesicular acetylcholine transporter (VAChT;

Eiden, 1998). ACh is then stored in the vesicles until it is released on demand (Wessler et al., 2003).

There are two types of receptors mediating cholinergic transmission with the ENS. ACh binds to nicotinic (nAChRs) and muscarinic receptors (mAChRs). nAChR are ligand- gated ion channels, whereas mAChR are G-protein-coupled receptors (Caulfield &

Birdsall, 1998). Thus, ACh binding generates variable postsynaptic potentials depending

on the receptor present on the cell membrane, with nAChRs mediating rapid excitatory

transmission and mAChRs mediating slow excitatory transmission (Harrington et al.,

2010). At the cholinergic synapse both classes of receptors are present on effector cells

(post- synaptic receptors) or on nerve terminals (pre-synaptic receptors) where they act

as autoreceptors regulating release of ACh from nerve terminals. Within the ENS

nAChRs are required for rapid neurotransmission, in order to propagate reflexes quickly

and produce fast responses to stimuli (Galligan, 2002). nAChR activation is the

predominant mechanism for cholinergic neurotransmission in enteric ascending reflex

pathways. mAChR mediate responses to ACh by activating second messenger cascades

and intracellular signalling pathways (Caulfield & Birdsall, 1998). The importance of

mAChR to determining the multiple roles ACh has in intestinal physiology is reflected in

their diverse distribution and different subtypes coupling to various G-proteins, thus

(34)

33

eliciting variable intracellular responses upon activation. ACh binding to mAChR either depolarizes the cell membrane, resulting in the initiation of another action potential, or hyperpolarizes the cell membrane, inhibiting additional action potentials.

1.4.2 Tachykinergic Neurotransmission

Neuropeptides are small molecules used by neurons for communication. However, they are not only important for neurotransmission, but they also have effects on tissue growth and differentiation, inflammation, immunomodulation and tumor growth. The production of neuropeptides occurs in the cell body of the neurons, and they are then transported to the varicosities and are released after stimulation.

A neuropeptide frequently discussed in inflammatory situations, including those of the intestine, is substance P (SP), a tachykinin expressed throughout the nervous and immune systems, that regulates an extraordinarily diverse range of physiological processes. This peptide consists of 11 amino acids and belongs to the tachykinin family of peptides, interacts with three neurokinin receptors (NKRs) encoded by three Tacr genes. SP has been discovered in extracts of horse brain and intestine with effects on intestinal contractility and blood pressure. The finding of SP was the first discovery of many other

“brain-gut neuropeptides,” which are present in enteric neurons and enteroendocrine cells as well as in neurons of the brain (Euler & Gaddum, 1931).

The preferred receptor for SP is the neurokinin-1 receptor (NK-1R) (Nakanishi, 1991;

Regoli & Nantel, 1991), but SP can also bind to NK-2R with low affinity (Hershey &

Krause, 1990). NK-1R is a member of the superfamily of guanin nucleotide binding- coupled receptors with seven membrane-spanning domains, three extracellular and intracellular loops, and extracellular NH2 and intracellular COOH termini, that couple with G-proteins to promote high-affinity binding and signal transduction (Hershey &

Krause, 1990). Binding of SP to the NK-1R mediates rapid endocytosis and internalization of the receptor (O'Connor et al., 2004).

SP, classically, is a peptide produced in sensory neurons, is a pain mediator and is involved in vaso-regulation and so-called neurogenic inflammation (Foreman, 1987;

Gamse et al., 1987). SP is also involved in immunomodulatory activities and has long

been considered to play a key role in IBD (Gross & Pothoulakis, 2007). SP has profound

effects for intestinal physiology and it is involved in the regulation of motility and

Riferimenti

Documenti correlati

Use of all other works requires consent of the right holder (author or publisher) if not exempted from copyright protection by the applicable

L’esperienza tragica della Grande guerra che travolse milioni di vite di uomini e donne sui diversi fronti è affrontata nel libro promosso dalla Fondazione Don Primo

Moreover, alteration of sequences including the TATA box (mutant LS2), and the region surrounding the transcription start site (mutant LS1) results in a substantial reduction of

The objective of this work is to propose and test a set of solutions to estimate a target optical feature at a given date from images acquired at adjacent dates, or even from

Starnini, ha permesso di procedere a una nuova documentazione grafica dei reperti e soprattutto ha comportato “una riconsiderazione critica sia della tipologia, sia della

Il 25 giugno raggiunsero lo Stretto di Hudson presso la punta settentrionale della Penisola del Labrador; seguendo la costa meridionale dello stretto, il 2 agosto

Sono stati valutati: la degranulazione dei mastociti, la concentrazione di istamina nel mezzo di coltura, la vasodilatazione, lo spessore epidermico, la proliferazione e i

Cellulose micro- and nanocrystals lead to a decrease in the adhesive strength of AQ500 related to the filler content, but for CMC-filled adhesive joints, this reduction in τ B